EP3296294A1 - Compound for treating or preventing breast cancer - Google Patents

Compound for treating or preventing breast cancer Download PDF

Info

Publication number
EP3296294A1
EP3296294A1 EP16792124.6A EP16792124A EP3296294A1 EP 3296294 A1 EP3296294 A1 EP 3296294A1 EP 16792124 A EP16792124 A EP 16792124A EP 3296294 A1 EP3296294 A1 EP 3296294A1
Authority
EP
European Patent Office
Prior art keywords
compound
alkoxy
compounds
fluorobenzoselenazole
breast cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP16792124.6A
Other languages
German (de)
French (fr)
Other versions
EP3296294A4 (en
EP3296294B1 (en
Inventor
Dongfang Shi
Changjin FU
Xi CHENG
Jianghua Zhu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jiangsu Atom Bioscience and Pharmaceutical Co Ltd
Original Assignee
Jiangsu Atom Bioscience and Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Atom Bioscience and Pharmaceutical Co Ltd filed Critical Jiangsu Atom Bioscience and Pharmaceutical Co Ltd
Priority to PL16792124T priority Critical patent/PL3296294T3/en
Publication of EP3296294A1 publication Critical patent/EP3296294A1/en
Publication of EP3296294A4 publication Critical patent/EP3296294A4/en
Application granted granted Critical
Publication of EP3296294B1 publication Critical patent/EP3296294B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D293/00Heterocyclic compounds containing rings having nitrogen and selenium or nitrogen and tellurium, with or without oxygen or sulfur atoms, as the ring hetero atoms
    • C07D293/10Heterocyclic compounds containing rings having nitrogen and selenium or nitrogen and tellurium, with or without oxygen or sulfur atoms, as the ring hetero atoms condensed with carbocyclic rings or ring systems
    • C07D293/12Selenazoles; Hydrogenated selenazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention belongs to the field of pharmaceutical chemistry.
  • the present invention particularly relates to 2-phenyl benzoselenazole compounds, methods for preparing the compounds, pharmaceutically acceptable salts thereof, prodrugs thereof or pharmaceutical compositions containing the compounds, as applications in preparing medicines for treating or preventing breast cancer of mammals.
  • breast diseases mainly include cyclomastopathy, breast fibroadenoma (benign tumor), and breast cancer (MengQingchun, TianYunxia et al., Survey and Analysis of Women Breast Diseases in Shijiazhuang, Hebei Medical Journal, 2012, 34(6):917-919 ).
  • Female breast cancer has become the world's second most common malignant tumor, a serious threat to women's health.
  • 2012 there were 1.67 million new women breast cancer cases worldwide, which accounted for 25.2% of all malignancies in females; there were 520,000 breast cancer deaths, which accounted for about 14.7% of all malignant deaths in females.
  • the breast cancer treatment includes operative treatment, radiation therapy and adjuvant chemotherapy.
  • the operative treatment is still main means for treating early-stage breast cancer.
  • the radiation therapy for the breast cancer is one of important measures for controlling local recurrence after the surgery, and the radiation therapy following the conservative surgery can significantly decrease the local recurrence rate by 75% averagely ( Lim M., Bellon J. R., Gelman R., et al, A prospective study of conservative surgery without radiation therapy in select patients with stage I breast cancer, Int. J. Radiat., Oncol. Biol. Phys., 2006, 65(4):1149 ).
  • CMF cyclophosphamide/methotrexate/fluorouracil
  • CAF cyclophosphamide/adriamycin/fluorouracil
  • FEC and the like are used clinically ( Bonadonna G., Brusamolino E., et al, Combination chemotherapy as an adjuvant treatment in operable breast cancer, N. Engl. J. Med., 1976, 294(8):405-410 ).
  • Taxanes such as paclitaxel (Taxol)
  • Taxol paclitaxel
  • the mechanism of taxol is that it acts on microtubule system, promotes tubulin polymerization, inhibits depolymerization, and cell cycle migration is blocked in M phase.
  • the selectivity of taxanes is poor, and there are serious side effects of bone marrow suppression, neurotoxicity, cardiovascular and liver toxicity, allergic reactions, which bring great physical and mental pain to patients.
  • Luminal subtype breast cancer estradiosarcoma
  • progesterone receptor positive Luminal subtype breast cancer
  • Hessaitin and Lapatinib are highly specific monoclonal antibodies for advanced breast cancer patients with HER2 overexpression; bevacizumab is also effective for taxane resistant advanced breast cancer( Miller K. D., Chap L. I., Holmes F. A., et al, Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer, J. Clin.
  • Phortress from this group of compounds had entered into phase I clinical study, but the toxicity of the compound to the liver and lungs, and the inability to determine the optimal dose of the treatment terminated the clinical trial. This may be due to the toxicity and defect of the compound itself, and this kind of compound has not been studied further. Currently, Phortress has not been further investigated as an anti-breast cancer drug in clinical use.
  • An objective of the present invention is to provide novel 2-phenyl benzoselenazole compounds on the basis of the prior art.
  • the compounds have excellent inhibition effect on the growth of breast cancer cell lines, but no inhibitory effect on the growth of some cell lines except breast cancer cell lines. These compounds have good selectivity. This series of compounds may become a new generation of drugs with high selectivity and low toxicity for the treatment of breast cancer.
  • R 1 and R 2 are independently selected from H, D, F, Cl,-CN, -CH 3 , -CF 3 , -OCH 3 , -OCH 2 CH 3 , -OCF 3 or -OCHF 2 .
  • R 1 and R 2 are independently selected from H, D, F, Cl,-CN, -CH 3 , -CF 3 , -OCH 3 or -OCH 2 CH 3 .
  • R 3 and R 4 are independently selected from H, D, halogen,-OH, -CN, -NH 2 , -CH 3 , -CH 2 CH 3 , -CF 3 , -OCH 3 , -OCH 2 CH 3 , -OCHF 2 or -OCF 3 .
  • R 5 is selected from H, -NH 2 , -CH 3 , -CF 3 , -OCH 3 , -OCHF 2 ,-OCF 3 , -OCH 2 CH 3 or -OCH 2 CF 3 .
  • R 1 and R 2 are independently selected from H, D, F, Cl, -CN, -CH 3 , -CF 3 or -CHF 2 ; and R 3 and R 4 are independently selected from H, D, F, Cl, Br, I, -CN, -CH 3 , -CF 3 , -OCH 3 , -OCH 2 CH 3 , -OCHF 2 or -OCF 3 .
  • the present invention further provides the following specific compounds, including pharmaceutically acceptable salts thereof orprodrugs thereof:
  • the compounds of the present invention may be prepared by the following method:
  • Corresponding diazoniumtetrafluoroborate (IB) was generated by the diazotization reaction between 2-nitroaniline compounds of formula (IA) and isoamyl nitrite in the presence of boron trifluoride diethyl etherate, the diazoniumtetrafluoroborate was then reacted with potassium selenocyanate to obtain phenyl selenocyanate compounds (IC), and the phenyl selenocyanate compounds were reacted in sodium/ethanol to obtain diselenium nitro compounds (ID). The compounds (ID) were reduced under certain conditions to obtain corresponding amino compounds, and the amino compounds were cyclized with corresponding benzaldehyde, benzoyl chloride or benzoic acid.
  • the obtained benzoselenazole compounds may be end products, or may be subject to a reduction reaction, a halogenation reaction or other reactions to obtain corresponding target products (I).
  • the groups R 1 , R 2 , R 3 , R 4 and R 5 are as defined above.
  • the present invention further provides pharmaceutical compositions containing any one of the compounds of the present invention, pharmaceutically acceptable salts thereof or easily hydrolysable prodrugs thereof as active components or main active components, supplemented by pharmaceutically acceptable adjuvants.
  • the compounds of the present invention, pharmaceutically acceptable salts thereof or easily hydrolysable prodrugs thereof can be applied in the preparation of medicines for treating or preventing breast cancer.
  • Hydrogen refers to protium (1H) which is a main stable isotope of hydrogen.
  • Deuterium is a stable isotope of hydrogen and also referred to as heavy hydrogen, and its symbol is D.
  • Halogen refers to fluorine atom, chlorine atom, bromine atom or iodine atom.
  • Haldroxyl refers to -OH.
  • Cyano refers to -CN.
  • Niro refers to -NO 2 .
  • Alkyl is a saturated aliphatic group having 1 to 10 carbon atoms, including a straight-chain group and a branched-chain group (the numerical range (e.g., 1 to 10) mentioned in the present application means that this group (alkyl in this case) may contain one carbon atom, two carbon atoms, three carbon atoms or even ten carbon atoms).
  • An alkyl containing 1 to 4 carton atoms is a low-level alkyl.
  • a low-level alkyl without any substituent group is an unsubstituted low-level alkyl, for example, methyl, ethyl, propyl, 2-propyl, n-butyl, isobutyl, tert-butyl or the like.
  • the alkyl may be substituted or unsubstituted.
  • Alkoxy represents -O- (unsubstituted alkyl) and -O- (unsubstitutedalkoxy), and further represents -O- (unsubstituted alkyl).
  • Representative embodiments include but are not limited to methoxy, ethoxy, propoxy, cyclopropoxy or the like.
  • “Pharmaceutically acceptable salts” include salts formed by the compounds of formula (I) with organic acids or inorganic acids, and represent salts maintaining the bioavailability and properties of the precursor compounds.
  • “Pharmaceutical compositions” refer to mixtures of one or more of the compounds described herein or pharmaceutically acceptable salts thereof and prodrugs thereof with other chemical components, for example, pharmaceutically acceptable carriers and excipients.
  • the pharmaceutical compositions are aimed at facilitating the administration of the compounds to a living body.
  • Prodrugs refer to compounds which have pharmacological actiononly after they are transformed into active compounds.
  • the prodrugs themselves have no or low bioactivity, and will become active substances after in vivo metabolism. This process is to increase the bioavailability of medicines, enhance the targeting performance and reduce the toxicity and side effects of medicines.
  • the present invention further claims pharmaceutical compositions containing any one of the above-described compounds, pharmaceutically acceptable salts thereof or easily hydrolysable prodrug amides thereof and other pharmaceutically active components.
  • the present invention also encompasses any one of the above-described compounds, pharmaceutically acceptable salts thereof, easily hydrolysable prodrug amides thereof or isomers thereof.
  • the present invention may be prepared into any clinically or pharmaceutically acceptable dosage form by the known methods in the art.
  • the present invention may be prepared into conventional solid preparations such as tablets, capsules, pills or granules, or oral liquid preparations such as oral solution, oral suspension or syrup.
  • proper filling agents, binding agents, disintegrating agents, lubricating agents and the like may be added.
  • the present invention may be prepared into injection preparations such as injection solution, sterile powder for injection and concentrated solution for injection.
  • existing conventional methods in the pharmaceutical field may be used.
  • no or proper additives may be added, depending upon the properties of the medicines.
  • the compounds of the present invention have a novel 2-phenyl-benzoselenazole matrix-cycle structure, and provide new mechanisms and treatment plans for the treatment or prevention of the breast cancer.
  • the compounds of the present invention have the following characteristics:
  • Step A a solution of 2-nitroaniline(15.0 g, 108 mmol) in dichloromethane (150 mL) was added dropwise to boron trifluoride diethyl etherate (23.1 g, 163 mmol) at -10°C to -15°C and stirred for 15 min, and a solution of isoamyl nitrite (15.26 g, 130 mmol) in dichloromethane (75 mL) was added dropwise at this temperature. At the end of addition, the reaction mixture was continuously stirred for 30 minutes, and then stirred for 30 minutes at -10°C to 0°C. The reaction system was added with cold petroleum ether (250 mL) dropwise and then filtered.
  • Step B a solution of potassium selenocyanate (8.0 g, 55.5 mmol) in water (80 mL) was added into a mixture of compound 1 (13.0 g, 54. 9 mmol) and water (300 mL) in an ice-water bath, and the reaction mixture was continuously stirred for 30 min at the end of addition. The reaction mixture was filtered, and the filter cake was washed with a small amount of water and then dried in vacuum at 60°C to obtain 1-nitro-2-phenyl selenocyanate ( 2 ) (11.2 g). The yield was 89.8%.
  • Step C sodium (6.0 g, 261 mmol) was added to a mixture of compound 2 (10.5 g, 46.2 mol) and absolute ethyl alcohol (520 mL) at room temperature, and the mixture was stirred for 1 h in a water bath.
  • the reaction mixture was cooled to 0°C to 5°C and filtered, the filter cake was washed with a small amount of cold ethanol, and the collected solid was suspended in methylbenzene (100 mL), heated to reflux to dissolve the product, and filtered immediately.
  • the filtrate was cooled to 0°C to 5°C to separate out solid and then filtered, and the filter cake was collected to obtain 1,2-di(2-nitrophenyl)diselenide ( 3 ) (4.5 g).
  • the yield was 48.4%.
  • Step D zinc powder (13.5 g, 206 mmol) was added into a suspension solution of compound 3 (4.5 g, 11.2 mmol) in acetic acid (90 mL) at 40°C, and the reaction mixture was heated to 100°C and continuously stirred for 2 h.
  • the reaction mixture was cooled below 50°C, 6M hydrochloric acid (40 mL) was slowly added dropwise and the mixture was filtered to remove insoluble substances.
  • the filtrate was adjusted with 20% sodium acetate aqueous solution until the pH value was 2 to 3, and the solid was separated out.
  • the reaction mixture was filtered, and the filter cake was dried to obtain di[(2-aminophenyl)seleno]zinc ( 4 ) (3.0 g). The yield was 77.8%.
  • Step E a mixture of compound 4 (3.6 g, 17.4 mmol) and 4-nitrobenzoyl chloride (4.77 g, 25.7 mmol) was stirred for 2 h at 110°C. The mixture was cooled to room temperature, saturated sodium bicarbonate aqueous solution (60 mL) was added, the mixture was extracted with ethyl acetate (50 mL ⁇ 3), and the combined organic phases were washed with saturated saline solution (20 mL) and dried over anhydrous sodium sulfate.
  • Step F compound 5 (450 mg, 1.48 mmol) was dissolved into ethanol (10 mL), 2M hydrochloric acid (15 mL) and tin powder (2.25 g, 6.74 mmol) were then added, and the mixture was refluxed and stirred for 2 h. Most of the solvent was evaporated under reduced pressure, then water (15 mL) was added, and the mixture was adjusted with dilute sodium hydroxide solution until the pH value was 9 to 10, extracted with dichloromethane (20 mL ⁇ 3), and dried over anhydrous sodium sulfate.
  • Step G a solution of NBS (117 mg, 0.657 mmol) in dichloromethane (15 mL) was added dropwise into a solution of compound 6 (200 mg, 0.732 mmol) in dichloromethane (5 mL) at -10°C, and the mixture was continuously stirred for 0.5 h at this temperature at the end of addition. The reaction mixture was washed with water (10 mL) and dried over anhydrous sodium sulfate.
  • Step A a solution of 2-nitro-4-fluoroaniline (5.0 g, 32.0 mmol) in dichloromethane (50 mL) was added dropwise to boron trifluoride diethyl etherate (6.8 g, 47.9 mmol) at -10°C to -15°C and stirred for 15 min, and a solution of isoamyl nitrite (4.5 g, 38.4 mmol) in dichloromethane (25 mL) was added dropwise at this temperature. At the end of addition, the mixture was continuously stirred for 30 minutes, and then stirred for 30 minutes at -10°C to 0°C. The reaction mixture was added with cooled petroleum ether (80 mL) dropwise and then filtered.
  • Step B a solution of potassium selenocyanate (4.84 g, 33.6 mmol) in water (30 mL) was added to a mixture of crude compound 8 (13.0 g) and water (170 mL) in an ice-water bath, and the reaction mixture was continuously stirred for 20 min at the end of addition. The reaction mixture was filtered, and the filter cake was washed with a small amount of water and then dried in vacuum at 60°C to obtain 4-fluoro-2-nitro-1-phenyl selenocyanate ( 9 ) (9.1 g). The compound was directly used in the next reaction without purification.
  • Step C sodium (4.1 g, 178 mmol) was added to a mixture of crude compound 9 (9.1 g) and absolute ethyl alcohol (300 mL) at room temperature, and the mixture was stirred for 1 h in a water bath. The mixture was cooled to 0°C to 5°C and filtered, and the filter cake was washed with a small amount of cold ethanol. The collected solid was suspended in methylbenzene (80 mL), heated to reflux to dissolve the product, and filtered immediately.
  • Step D compound 10 (1.49 g, 3.4 mmol) and Raney nickel (1.2 g) were suspended in isopropanol (30 mL), 85% hydrazine hydrate (1.8 mL) was added, and the mixture was heated to reflux and continuously stirred for 2.5 h. The mixture was immediately filtered through a celite pad, the filter cake was washed with a small amount of isopropanol, and the filtrate was collected.
  • Step E a mixture containing compound 11 (370 mg, 0.978 mmol), 4-nitrobenzaldehyde (296 mg, 1.958 mmol), sodium metabisulfite (372 mg, 1.956 mmol) and anhydrous DMSO (15 mL) was stirred for 72 h under nitrogen at 120°C. The mixture was cooled to room temperature, added with saturated ammonium chloride aqueous solution (60 mL), extracted with ethyl acetate (25 mL ⁇ 3), and dried over anhydrous sodium sulfate.
  • Step F compound 12 (70 mg, 0.218 mmol) was dissolved into ethanol (5 mL) and then added with stannous chloride hydrate (246 mg, 1.09 mmol), and the mixture was refluxed and stirred for 5 h. The mixture was cooled to room temperature, saturated saline solution (20 mL) was added and the mixture was extracted with ethyl acetate (25 mL ⁇ 3), and dried over anhydrous sodium sulfate.
  • Step G a solution of NBS (29.6 mg, 0.166 mmol) in dichloromethane (40 mL) was added dropwise to a solution of compound 13 (44 mg, 0.151 mmol) in dichloromethane (40 mL) in an ice-salt bath, and the mixture was continuously stirred for 10 min at this temperature at the end of addition.
  • the reaction mixture was washed with water (20 mL ⁇ 2) and dried over anhydrous sodium sulfate.
  • Step A zinc powder (5.5 g, 84.1 mmol) was added to a suspension of compound 10 (2.0 g, 4.57 mmol) in acetic acid (40 mL) at 40°C, then heated to 100°C and continuously stirred for 3 h.
  • the reaction mixture was cooled below 50°C, 6M hydrochloric acid (40 mL) was slowly added and the mixture was filtered to remove insoluble substances.
  • the filtrate was adjusted with 20% sodium acetate until the pH value was 2 to 3, and the solid was collected by filtration and dried to obtain di[(2-amino-4-fluorophenyl)seleno]zinc ( 15 ) (1.2 g). The yield was 61.9%.
  • Step B a mixture of compound 15 (590 mg, 3.10 mmol) and 3-methyl-4-nitrobenzoyl chloride (663 mg, 3.32 mmol) was stirred for 4 h at 100°C. The mixture was cooled to room temperature, saturated sodium bicarbonate aqueous solution (15 mL) was added and the mixture was extracted with ethyl acetate (30 mL ⁇ 2). The combined organic phases were washed with saturated saline solution (10 mL) and dried over anhydrous sodium sulfate.
  • Step C compound 16 (200 mg, 0.597 mmol) was dissolved into ethanol (5 mL), 3M hydrochloric acid (4 mL) and tin powder (800 mg, 6.74 mmol) were then added and the mixture was refluxed and stirred for 1.5 h. Most of the solvent was evaporated under reduced pressure, water (15 mL) was then added and the mixture was adjusted with dilute sodium hydroxide solution until the pH value was 9 to 10, extracted with ethyl acetate (20 mL ⁇ 2), and dried over anhydrous sodium sulfate.
  • NBS (57 mg, 0.320 mmol) was added to a solution of compound 17 (88 mg, 0.229 mmol) in DMF (5 mL) at room temperature, and the mixture was continuously stirred for 20 min at this temperature at the end of addition.
  • Water 25 mL was added to the mixture which was then extracted with ethyl acetate (20 mL ⁇ 2), and the combined organic phases were washed with saturated sodium bicarbonate aqueous solution (10 mL) and dried over anhydrous sodium sulfate.
  • Steps A, B and C referred to steps A, B and C in Embodiment 2.
  • Step D compound 21 (1.9 g, 3.393 mmol) was dissolved into ethanol (40 mL) and stannous chloride hydrate (3.8 g, 16.84 mmol) was then added, and the mixture was refluxed and stirred for 3.5 h under nitrogen. The mixture was cooled to room temperature, saturated saline solution (20 mL) was added, the mixture was extracted with ethyl acetate (25 mL ⁇ 3), and dried over anhydrous sodium sulfate.
  • Step E referring to step E in Embodiment 2, to get 5-bromo-2-(4-nitrophenyl)benzoselenazole ( 23 ).
  • Steps F and G referring to step F in Embodiment 2 to get compound 24.
  • the compound 23 was reduced to obtain a compound 24.
  • Compound 24 (80 mg, 0.227 mmol) was dissolved into DMF (5 mL), NCS (34 mg, 0.255 mmol) was added and the mixture was stirred overnight at room temperature.
  • Water 25 mL was added to the mixture which was extracted with ethyl acetate (15 mL ⁇ 3), and the combined organic phases were washed with saturated sodium bicarbonate aqueous solution (10 mL) and dried over anhydrous sodium sulfate.
  • the method for preparing compound 26 refers to steps A, B, C and D in Embodiment 2 and steps E and F in Embodiment 5.
  • 2-Nitro-4-fluoroaniline in step A of Embodiment 2 was replaced with 2-nitro-4-methylaniline
  • 4-nitrobenzaldehyde in step E of Embodiment 5 was replaced with 3-methyl-4-nitrobenzaldehyde.
  • the method for preparing compound 27 refers to steps A, B, C and D in Embodiment 2 and steps E and F in Embodiment 5.
  • 2-nitro-4-fluoroaniline in step A of Embodiment 2 was replaced with 2-nitro-4-trifluoromethylaniline
  • 4-nitrobenzaldehyde in step E of Embodiment 5 was replaced with 3-methyl-4-nitrobenzaldehyde.
  • the method for preparing compound 28 refers to step B in Embodiment 3, wherein acyl chloride in the reaction equation was prepared by reacting a corresponding acid with thionyl chloride.
  • the method for preparing compound 29 refers to steps A, B, C and D in Embodiment 2 and steps E and F in Embodiment 5.
  • 2-nitro-4-fluoroaniline in step A was replaced with 2-nitro-5-fluoroaniline
  • 3-methyl-4-nitrobenzaldehyde in step E of Embodiment 5 was replaced with 3-methyl-4-nitrobenzaldehyde.
  • the solvent was ethanol so that F at the sixth site of benzaldehyde was substituted with ethoxy.
  • the final resulting product was 4-(6-ethoxybenzoselenazole-2-yl)-2-methylaniline ( 29 ).
  • the method for preparing compound 30 refers to steps A, B, C and D in Embodiment 2 and steps E and F in Embodiment 5.
  • 2-nitro-4-fluoroaniline in step A of Embodiment 2 was replaced with 2-nitro-4,5-difluoroaniline
  • 4-nitrobenzaldehyde in step E of Embodiment 5 was replaced with 3-methyl-4-nitrobenzaldehyde.
  • the solvent was ethanol so that F at the sixth site of benzaldehyde was substituted with ethoxy.
  • the final resulting product was 4-(6-ethoxy-5-fluorobenzoselenazole-2-yl)-2-methylaniline ( 30 ).
  • step D in Embodiment 5 and step E in Embodiment 2 compound 32 was synthesized as the aimed product while using compound 3 and 4-nitrobenzaldehyde as the starting materials.
  • the compound 3 was used as raw material, and the method for preparing the compound 32 refers to step D in Embodiment 5 and step E in Embodiment 2.
  • 4-nitrobenzaldehyde in step E of Embodiment 2 was replaced with 3-hydroxyl-4-methoxybenzaldehyde.
  • Step A tributylphosphine (1.28 g, 6.33 mmol) was added to a solution of compound 11 (800 mg, 2.12 mmol) in methylbenzene (15 mL), then stirred for 5 min under nitrogen. To the mixture was added 3-fluoro-4-nitrobenzoic acid (392 mg, 2.12 mmol), and the resulting mixture was refluxed for 48 h under nitrogen. The mixture was cooled to room temperature, water (25 mL) was added and the mixture was adjusted with saturated sodium carbonate solution until the pH value was 9 to 10.
  • Step B 2-bromo-6-fluoro-(5-fluorobenzoselenazole-2-yl)aniline ( 35 ) was prepared by using compound 34 as raw material, and the method refers to step G in Embodiment 2.
  • Step A a solution of compound 11 (800 mg, 2.12 mmol) and tributylphosphine (1.28 g, 6.32 mmol) in methylbenzene (15 mL) was stirred for 5 min under nitrogen, 3-nitro-4-methylbenzoic acid (380 mg, 2.09 mmol) was added, and the mixture was refluxed and stirred for 48 h under nitrogen. The mixture was cooled to room temperature, water (30 mL) was added and the mixture was adjusted with 2M sodium hydroxide solution until the pH value was 9 to 10. The mixture was extracted with ethyl acetate (20 mL ⁇ 3) and dried over anhydrous sodium sulfate.
  • Step B 5-(5-fluorobenzoselenazole-2-yl)-2-methylaniline ( 37 ) was prepared by using compound 36 as raw material, and the experimental operation refers to step F in Embodiment 2.
  • the method for preparing compound 38 refers to step E in Embodiment 2, wherein 4-nitrobenzaldehyde in step E of Embodiment 2 was replaced with 3-chloro-4(trifluoromethoxy)benzaldehyde.
  • 1 H NMR (DMSO- d 6 , 300 MHz) ⁇ 8.35 (d, J 1.2 Hz, 1H), 8.30-8.27 (m, 1H), 8.17-8.15 (m, 1H), 7.98-7.96 (m, 1H), 7.77-7.75 (m, 1H), 7.38-7.34 (m, 1H).
  • Step A compound 22 and 3-methyl-4-nitrobenzaldehyde were used as raw materials, and the experimental operation of preparing compound 39 refers to step E in Embodiment 2.
  • Step B compound 39 (42 mg, 0.106 mmol) was suspended in DMF (5 mL) and deuteroxide (0.5 mL) and 5% palladium carbon were then added, and the mixture was stirred overnight under deuterium gas under normal pressure. The mixture was filtered through a celite pad, water (20 mL) was added and the mixture was extracted with ethyl acetate (20 mL ⁇ 2), and the combined organic phases were washed with saturated saline solution (10 mL ⁇ 2) and dried over anhydrous sodium sulfate.
  • the method for preparing compound 41 refers to step A in Embodiment 13, wherein 3-fluoro-4-nitrobenzaldehyde in step A of Embodiment 13 was replaced with 3,5-difluoro-4-nitrobenzaldehyde.
  • Step A a solution of NBS (5.0 g, 28.1 mmol) in DMF (20 mL) was added dropwise to a solution of 2-fluoro-6-methylaniline (3.5 g, 28.0 mmol) in DMF (10 mL) in an ice-water bath, and the mixture was continuously stirred for 5 min at the end of addition. The ice-water bath was removed, and the reaction mixture was stirred 0.5 h at room temperature.
  • Step B a mixture of compound 42 (5.3 g, 26.0 mmol), cuprous cyanide (3.0 g, 33.5 mmol) and N-methylpyrrolidone (15 mL) was stirred overnight under nitrogen at 180°C. Water (75 mL) was added to the mixture which was extracted with ethyl acetate (50 mL ⁇ 3), and the combined organic phases were successively washed with water (30 mL ⁇ 2) and saturated saline solution (30 mL) and dried over anhydrous sodium sulfate.
  • Step C a mixture of compound 43 (2.95 g, 19.6 mmol), 1M sodium hydroxide solution (50 mL) and ethanol (5 mL) was refluxed and stirred overnight. The mixture was cooled to room temperature, water (50 mL) was added and the mixture was washed with MTBE (20 mL ⁇ 2), and the water phase was used as the product. The water phase was adjusted with 2M hydrochloric acid until the pH value was 3 to 4, and the solid was separated out. The mixture was filtered, and the filter cake was dried to obtain 4-amino-3-fluoro-5-methylbenzoic acid ( 44 ) (2.90 g). The yield was 87.5%.
  • Step D tributylphosphine (2.15 g, 10.6 mmol) was added to a mixture of compound 11 (1.74 g, 4.60 mmol), compound 44 (600 mg, 3.55 mmol) and methylbenzene (25 mL), and the mixture was refluxed and stirred for 48 h under nitrogen. The mixture was cooled to room temperature, water (40 mL) was added and the mixture was adjusted with 2M sodium hydroxide solution until the pH value was 9 to 10. The mixture was extracted with ethyl acetate (40 mL ⁇ 3), and the combined organic phases were washed with saturated saline solution (25 mL) and dried over anhydrous sodium sulfate.

Abstract

Disclosed are a class of compounds for treating or preventing breast cancer, in particular 2-phenyl benzoselenazole compounds, and a pharmaceutically acceptable salt thereof and an easily hydrolysable prodrug thereof. Further disclosed are a pharmaceutical composition containing these compounds and the use of such compounds for for treating or preventing breast cancer in mammals. The compounds of the present invention can effectively inhibit or reduce the growth or proliferation of mammalian breast cancer cells, and at the same time, such compounds do not inhibit the growth of partial test cells, apart from breast cancer cells in mammals, and have a good selectivity. Such compounds show a more obvious medicinal effect, and have high selectivivity, low toxicity and few side effects.

Description

    Technical Field of the Invention
  • The present invention belongs to the field of pharmaceutical chemistry. The present invention particularly relates to 2-phenyl benzoselenazole compounds, methods for preparing the compounds, pharmaceutically acceptable salts thereof, prodrugs thereof or pharmaceutical compositions containing the compounds, as applications in preparing medicines for treating or preventing breast cancer of mammals.
  • Background of the Invention
  • As common diseases among women, breast diseases mainly include cyclomastopathy, breast fibroadenoma (benign tumor), and breast cancer (MengQingchun, TianYunxia et al., Survey and Analysis of Women Breast Diseases in Shijiazhuang, Hebei Medical Journal, 2012, 34(6):917-919). Female breast cancer has become the world's second most common malignant tumor, a serious threat to women's health. In 2012, there were 1.67 million new women breast cancer cases worldwide, which accounted for 25.2% of all malignancies in females; there were 520,000 breast cancer deaths, which accounted for about 14.7% of all malignant deaths in females. (Bernard W., Stewart, Christopher P., Wild World Cancer Report 2014, The international agency for research on cancer, World Health Organization.)The incidences of breast cancer have obvious geographical distribution differences. Europe and the United States have the highest incidences of breast cancer in the world, and the incidences are relatively low in Asia and Africa(Perera N. M.,andGui G. P., Multi-ethnic differences in breast cancer: current concepts and future directions, Int. J. Cancer, 2003, 106:463-467). Although the rates of breast cancer in Europe and the United States continue to rise, the mortality rates have decreased year by year.This is mainly due to early detection and treatment of the breast cancer patients. However, in Asia and Africa, due to poor detection techniques and treatment means, the morbidity and mortality of the breast cancer are on the rise (Kawamura T., and Sobue T., Comparison of breast cancer mortality in five countries: France, Italy, Japan, the UK and the USA from the WHO mortality database (1960-2000), Jpn. J. Clin. Oncol., 2005, 35(12):758-759). With the changes in the lifestyle, dietary habits, and environmental factors of Chinese females, the breast cancer becomes one of main factors threatening the health of Chinese females.
  • Clinically, based on the immunohistochemical techniques and according to the level of receptors (ER, PR and HER2) and the cell proliferation genetic markers (Ki-67), there are different subtypes of breast cancer: 1. triple-negative breast cancer (ER, PR and HER2 are all negative); 2.Luminal breast cancer (ER and PR are positive, but HER2 and Ki67 are different in expression); and3.HER2-overexpressioninbreast cancer (ER and PR deficiency, and HER-overexpression), wherein the triple-negative breast cancer accounts for about 10% to 20% of the breast cancer, which is highest in degree of malignancy and very easy to cause epithelial-mesenchymal transition, and has worse prognosis than other subtypes of breast cancer. Both endocrinotherapy and targeted therapies are ineffective for the triple-negative breast cancer (Tan A. R., and Swain S. M., Therapeutic strategies for triple-negative breast cancer, Cancer J., 2008, 14(6): 43-351).
  • At present, the breast cancer treatment includes operative treatment, radiation therapy and adjuvant chemotherapy. The operative treatment is still main means for treating early-stage breast cancer. The radiation therapy for the breast cancer is one of important measures for controlling local recurrence after the surgery, and the radiation therapy following the conservative surgery can significantly decrease the local recurrence rate by 75% averagely (Lim M., Bellon J. R., Gelman R., et al, A prospective study of conservative surgery without radiation therapy in select patients with stage I breast cancer, Int. J. Radiat., Oncol. Biol. Phys., 2006, 65(4):1149).
  • The chemotherapy, as a systematic adjuvant treatment method, has attracted extensive attention in the system therapy for the breast cancer. At the very beginning, CMF (cyclophosphamide/methotrexate/fluorouracil), CAF (cyclophosphamide/adriamycin/fluorouracil), FEC and the like are used clinically (Bonadonna G., Brusamolino E., et al, Combination chemotherapy as an adjuvant treatment in operable breast cancer, N. Engl. J. Med., 1976, 294(8):405-410). The emergence of taxanes, such as paclitaxel (Taxol), has greatly improved the survival rate of early breast cancer patients, and has provided help for the treatment of metastatic breast cancer. The mechanism of taxol is that it acts on microtubule system, promotes tubulin polymerization, inhibits depolymerization, and cell cycle migration is blocked in M phase. But the selectivity of taxanes is poor, and there are serious side effects of bone marrow suppression, neurotoxicity, cardiovascular and liver toxicity, allergic reactions, which bring great physical and mental pain to patients. Currently, endocrine therapy has been used as a standard adjuvant therapy for Luminal subtype breast cancer (estrogen receptor / progesterone receptor positive), which can reduce the annual mortality rate of this subtype of breast cancer by more than 31% (Gralow J. R., Burtein H. J., Wood W., Preoperative therapy in invasive breast cancer: pathologic assessment and systemic therapy issues in operable disease, J. Clin. Oncol., 2008, 26(5): 814-819).
  • Molecular targeted therapy is a hot spot in the field of breast cancer treatment, and many clinical trials have achieved good results in china. Among them, Hessaitin and Lapatinib are highly specific monoclonal antibodies for advanced breast cancer patients with HER2 overexpression; bevacizumab is also effective for taxane resistant advanced breast cancer(Miller K. D., Chap L. I., Holmes F. A., et al, Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer, J. Clin. Oncol., 2005, 23(4):792-799).However, for patients with triple-negativeand HER2-overexpression breast cancer, because of its estrogen receptor and progesterone receptor were negative, so to tamoxifen, letrozole, anastrozole, exemestane and other traditional endocrine therapy had no reaction. Therefore, it is urgent to develop targeted anti breast cancer agents according to different subtypes of breast cancer.
  • Compounds containing benzothiazole have extensive biological activities (Weekes A. A., and Westwell A. D., 2-Arylbenzothiazole as a privileged scaffold in drug discovery, Curr. Med. Chem., 2009, 16(19):2430-2440). Stevens et al. (WO0114354A1 ) had reported 2-phenyl benzoxazole compounds or 2-phenyl benzothiazoles and their derivatives thereof, which have a highly selective inhibitory activity against breast cancer cells.
    Figure imgb0001
  • Shi and Aiellohad reported a group of compounds with highly selective inhibitory against breast cancer cell lines(Shi D.-F., Bradshaw T. D., Wrigley S., et al, Antitumourbenzothiazoles. 3. Synthesis of 2-(4-aminophenyl)-benzothiazoles and evaluation of their activities against breast cancer cell lines in vitro and in vivo. [J]. J Med Chem. 1996, 39:3375-3384; Aiello S., Wells G., Stone E. L., et al, Synthesis and biological properties of benzothiazole, benzoxazole, and chromen-4-one analogues of the potent antitumor 2-(3,4-dimethoxyphenyl)-5-fluorobenzothiazole (PMX-610, NSC721648), J. Med. Chem., 2008, 51:5135-5139.). The mechanism of action is to induce the expression of CYP1A1 in the P450 enzyme system in the cell, and then metabolized by CYP1A1 to become a highly active substance, inducing tumor cell DNA damage and apoptosis(Bradshaw T. D., Stevens M. F. G., Westwell A. D., The discovery of the potent and selective antitumour agent 2-(4-amino- 3-methylphenyl)benzothiazole(DF203) and related compounds. Curr. Med. Chem., 2001, 8(2):203-210; Rodriguez M. And Potter D. A., CYP1A1 regulates breast cancer proliferation and surviral, Mol. Cancer Res. 2013, 11(7):780-792; Wang K. and Guengerich F. P., Bioactivation of fluorinated 2-arylbenzothiazole antitumor molecules by human cytochrome P450s 1A1 and 2W1 and deactivation by cytochrome P4502S1, Chem. Res. Toxicol., 2012, 25,1740-1751.). Phortress from this group of compounds had entered into phase I clinical study, but the toxicity of the compound to the liver and lungs, and the inability to determine the optimal dose of the treatment terminated the clinical trial. This may be due to the toxicity and defect of the compound itself, and this kind of compound has not been studied further. Currently, Phortress has not been further investigated as an anti-breast cancer drug in clinical use.
  • Akama et al had reported a series of 5,4'-diamino-6,8,3'-trifluoroflavone compounds, which have good anti-tumor activity, especially for breast cancer cell proliferation inhibition(Akama T., Ishida H., Kimura U., et al, Structure-actuvity relationships of the 7-substituents of 5,4'-diamino-6,8,3'-trifluoroflavone, a potent antitumor agent, J. Med. Chem. 1998, 41, (12):2056-2067).
    Figure imgb0002
  • Due to their applications in antitumor, antiviral therapy and nervous system related disease, selenium containing medicines have become a hotspot of the research and development of scholars both in China and foreign countries. The research has mainly focused on anti-tumor, anti-inflammatory, anti-hypertension and the like (Romualdo C., Stefania C., Marina D. G., et al, Novel selenium-containing non-natural diamino acids, Tetrahedron Lett., 2007, 48(7):1425-1427.). Among them, the role of organic selenium compounds in tumor prevention and treatment has long been widely studied. A large number of studies have shown that organic selenium compounds have definite effect on the resistance of a series of tumors, including colorectal cancer, tumors of digestive tract, respiratory tract cancer, skin cancer, lung cancer, colon cancer, prostate cancer, gastric cancer, liver cancer, breast cancer, ovarian cancer, etc.(El-bayoumy K., and Sinha R., Mechanisms of mammary cancer chemoprevention by organoselenium compounds, Mutat. Res., 2004(551): 181-197.).
  • Summary of the Invention
  • An objective of the present invention is to provide novel 2-phenyl benzoselenazole compounds on the basis of the prior art. The compounds have excellent inhibition effect on the growth of breast cancer cell lines, but no inhibitory effect on the growth of some cell lines except breast cancer cell lines. These compounds have good selectivity.This series of compounds may become a new generation of drugs with high selectivity and low toxicity for the treatment of breast cancer.
  • The objective of the present invention can be achieved by the following schemes. Compounds of the general formula (I), pharmaceutically acceptable salts thereof or prodrugs thereof are provided:
    Figure imgb0003
    wherein:
    • R1 and R2 are independently selected from H, D, halogen, -CN, C1-3 alkyl, substituted C1-3 alkyl, C1-3alkoxy or substituted C1-3alkoxy, and the substituent group is selected from D, halogen or C1-3alkoxy;
    • R3 and R4 are independently selected from H, D, halogen, -OH, -CN, -NH2, substituted -NH2, C1-3 alkyl, substituted C1-3 alkyl, C1-3alkoxy or substituted C1-3alkoxy, and the substituent group is selected from D, halogen, C1-3 alkyl or C1-3alkoxy; and
    • R5 is selected from H, -OH, -NH2, C1-3 alkyl, substituted C1-3 alkyl, C1-3alkoxy or substituted C1-3alkoxy, and the substituent group is selected from D, halogen, -OH, -NH2 or C1-3alkoxy.
  • In a preferred scheme:
    • R1 and R2 are independently selected from H, D, halogen, -CN, C1-3 alkyl, substituted C1-3 alkyl, C1-3alkoxy or substituted C1-3alkoxy, and the substituent group is selected from D, F or C1-3alkoxy;
    • R3 and R4 are independently selected from H, D, halogen, -OH, -CN, -NH2, substituted -NH2, C1-3 alkyl, substituted C1-3 alkyl, C1-3alkoxy or substituted C1-3alkoxy, and the substituent group is selected from D, F, C1-3 alkyl or C1-3alkoxy; and
    • R5 is selected from H, -OH, NH2, C1-3alkoxy or substituted C1-3alkoxy, and the substituent group is selected from D, F, -OH, -NH2 or C1-3alkoxy.
  • In a preferred scheme, R1 and R2 are independently selected from H, D, F, Cl,-CN, -CH3, -CF3, -OCH3, -OCH2CH3, -OCF3 or -OCHF2.
  • In a preferred scheme, R1 and R2 are independently selected from H, D, F, Cl,-CN, -CH3, -CF3, -OCH3 or -OCH2CH3.
  • In a preferred scheme, R3 and R4 are independently selected from H, D, halogen,-OH, -CN, -NH2, -CH3, -CH2CH3, -CF3, -OCH3, -OCH2CH3, -OCHF2 or -OCF3.
  • In a preferred scheme, R5 is selected from H, -NH2, -CH3, -CF3, -OCH3, -OCHF2,-OCF3, -OCH2CH3 or -OCH2CF3.
  • As a preferred scheme of the present invention, the compounds of formula (II), pharmaceutically acceptable salts thereof orprodrugs thereof are used:
    Figure imgb0004
    where R1 to R4 are as defined above.
  • In a preferred scheme, in the formula (II) or (I), R1 and R2 are independently selected from H, D, F, Cl, -CN, -CH3, -CF3 or -CHF2; and R3 and R4 are independently selected from H, D, F, Cl, Br, I, -CN, -CH3, -CF3, -OCH3, -OCH2CH3, -OCHF2 or -OCF3.
  • The present invention further provides the following specific compounds, including pharmaceutically acceptable salts thereof orprodrugs thereof:
    • 4-(benzoselenazole-2-yl)-2-bromoaniline,
    • 2-bromo-4-(5-fluorobenzoselenazole-2-yl)aniline,
    • 4-(5-fluorobenzoselenazole-2-yl)-2-methylaniline,
    • 4-(5-fluorobenzoselenazole-2-yl)aniline,
    • 2-bromo-4-(5-fluorobenzoselenazole-2-yl)-6-methylaniline,
    • 4-(5-bromobenzoselenazole-2-yl)-2-chloroaniline,
    • 2-methyl-4-(5-methylbenzoselenazole-2-yl)aniline,
    • 2-methyl-4-[5-(trifluoromethyl)benzoselenazole-2-yl]aniline,
    • 2-(3,4-dimethoxy-phenyl)-5-fluoro-benzoselenazole,
    • 4-(6-ethoxybenzoselenazole-2-yl)-2-methylaniline,
    • 4-(6-ethoxy-5-fluorobenzoselenazole-2-yl)-2-methylaniline,
    • 5-(benzoselenazole-2-yl)-2-methoxyphenol,
    • 2-(3,4-dimethoxyphenyl)benzoselenazole,
    • 2-fluoro-4-(5-fluorobenzoselenazole-2-yl)aniline,
    • 2-bromo-6-fluoro-4-(5-fluorobenzoselenazole-2-yl)aniline,
    • 5-(5-fluorobenzoselenazole-2-yl)-2-methylaniline,
    • 2-[3-chloro-4-(trifluoromethoxy)phenyl]-5-fluorobenzoselenazole,
    • 4-(5-deuterobenzoselenazole-2-yl)-2-methylaniline,
    • 2,6-difluoro-4-(5-fluorobenzoselenazole-2-yl)aniline, and
    • 2-fluoro-4-(5-fluorobenzoselenazole-2-yl)-6-methylaniline.
  • The compounds of the present invention may be prepared by the following method:
    Figure imgb0005
  • Corresponding diazoniumtetrafluoroborate (IB) was generated by the diazotization reaction between 2-nitroaniline compounds of formula (IA) and isoamyl nitrite in the presence of boron trifluoride diethyl etherate, the diazoniumtetrafluoroborate was then reacted with potassium selenocyanate to obtain phenyl selenocyanate compounds (IC), and the phenyl selenocyanate compounds were reacted in sodium/ethanol to obtain diselenium nitro compounds (ID). The compounds (ID) were reduced under certain conditions to obtain corresponding amino compounds, and the amino compounds were cyclized with corresponding benzaldehyde, benzoyl chloride or benzoic acid. The obtained benzoselenazole compounds may be end products, or may be subject to a reduction reaction, a halogenation reaction or other reactions to obtain corresponding target products (I). The groups R1, R2, R3, R4 and R5 are as defined above.
  • The present invention further provides pharmaceutical compositions containing any one of the compounds of the present invention, pharmaceutically acceptable salts thereof or easily hydrolysable prodrugs thereof as active components or main active components, supplemented by pharmaceutically acceptable adjuvants.
  • The compounds of the present invention, pharmaceutically acceptable salts thereof or easily hydrolysable prodrugs thereof can be applied in the preparation of medicines for treating or preventing breast cancer.
  • Unless otherwise stated, the terms in the claims and the description are defined as below.
  • "Hydrogen" refers to protium (1H) which is a main stable isotope of hydrogen. "Deuterium" is a stable isotope of hydrogen and also referred to as heavy hydrogen, and its symbol is D.
  • "Halogen" refers to fluorine atom, chlorine atom, bromine atom or iodine atom.
  • "Hydroxyl"refers to -OH.
  • "Cyano"refers to -CN.
  • "Nitro"refers to -NO2.
  • "Alkyl" is a saturated aliphatic group having 1 to 10 carbon atoms, including a straight-chain group and a branched-chain group (the numerical range (e.g., 1 to 10) mentioned in the present application means that this group (alkyl in this case) may contain one carbon atom, two carbon atoms, three carbon atoms or even ten carbon atoms). An alkyl containing 1 to 4 carton atoms is a low-level alkyl. A low-level alkyl without any substituent group is an unsubstituted low-level alkyl, for example, methyl, ethyl, propyl, 2-propyl, n-butyl, isobutyl, tert-butyl or the like. The alkyl may be substituted or unsubstituted.
  • "Alkoxy" represents -O- (unsubstituted alkyl) and -O- (unsubstitutedalkoxy), and further represents -O- (unsubstituted alkyl). Representative embodiments include but are not limited to methoxy, ethoxy, propoxy, cyclopropoxy or the like.
  • "Pharmaceutically acceptable salts"include salts formed by the compounds of formula (I) with organic acids or inorganic acids, and represent salts maintaining the bioavailability and properties of the precursor compounds.
  • These salts include:
    1. (1) salts formed by the compounds with acids, which are obtained by reacting free bases of the precursor compounds with inorganic acids or organic acids, wherein the inorganic acids include (but not limited to): for example, hydrochloric acid, hydrobromic acid, nitric acid, phosphoric acid, meta phosphoric acid, sulfuric acid, sulfurous acid, perchloric acid and the like; the organic acids include (but not limited to): for example, acetic acid, propanoic acid, acrylic acid, oxalic acid, (D) or (L) malic acid, fumaric acid, maleic acid, hydroxybenzoic acid, γ-hydroxybutyric acid, methoxybenzoic acid, phthalic acid, methanesulfonic acid, ethanesulfonic acid, 1-naphthalenesulphonic acid, 2-naphthalenesulphonic acid, p-toluenesulfonic acid, salicylic acid, tartaric acid, citric acid, lactic acid, mandelic acid, succinic acid, malonic acid and the like; and
    2. (2) salts generated by substituting acidic protons in the precursor compounds with metal ions or coordinating the acidic protons in the precursor compounds with organic bases, wherein the metal ions include, for example, alkali metal ions, alkaline-earth metal ions or aluminum ions; and the organic bases include, for example, ethanolamine, diethanolamine, triethanolamine, trometamol, N-methylglucamine and the like.
  • "Pharmaceutical compositions" refer to mixtures of one or more of the compounds described herein or pharmaceutically acceptable salts thereof and prodrugs thereof with other chemical components, for example, pharmaceutically acceptable carriers and excipients. The pharmaceutical compositions are aimed at facilitating the administration of the compounds to a living body.
  • "Prodrugs" refer to compounds which have pharmacological actiononly after they are transformed into active compounds. The prodrugs themselves have no or low bioactivity, and will become active substances after in vivo metabolism. This process is to increase the bioavailability of medicines, enhance the targeting performance and reduce the toxicity and side effects of medicines.
  • The present invention further claims pharmaceutical compositions containing any one of the above-described compounds, pharmaceutically acceptable salts thereof or easily hydrolysable prodrug amides thereof and other pharmaceutically active components.
  • The present invention also encompasses any one of the above-described compounds, pharmaceutically acceptable salts thereof, easily hydrolysable prodrug amides thereof or isomers thereof. The present invention may be prepared into any clinically or pharmaceutically acceptable dosage form by the known methods in the art. For oral administration, the present invention may be prepared into conventional solid preparations such as tablets, capsules, pills or granules, or oral liquid preparations such as oral solution, oral suspension or syrup. During the preparation of oral preparations, proper filling agents, binding agents, disintegrating agents, lubricating agents and the like may be added. For parenteral administration, the present invention may be prepared into injection preparations such as injection solution, sterile powder for injection and concentrated solution for injection. During the preparation of injection preparations, existing conventional methods in the pharmaceutical field may be used. During the preparation of injection preparations, no or proper additives may be added, depending upon the properties of the medicines.
  • The compounds of the present invention have a novel 2-phenyl-benzoselenazole matrix-cycle structure, and provide new mechanisms and treatment plans for the treatment or prevention of the breast cancer. The compounds of the present invention have the following characteristics:
    1. (1) the compounds of the present invention can effectively inhibitor reduce the growth or proliferation of breast cancer cells of mammals with tumors; and
    2. (2) the compounds of the present invention have better physicochemical properties, more remarkable pharmaceutical effect, low toxicity and low side effect.
    Detailed Description of the Invention
  • To make the objectives, technical schemes and advantages of the present invention clearer, the present invention will be further described below in details by specific implementations. It should be understood that the descriptions are merely exemplary and not intended to limit the scope of the present invention.
  • Synthesis embodiments Embodiment 1: synthesis of 4-(benzoselenazole-2-yl)-2-bromoaniline (7)
  • Figure imgb0006
  • Step A: a solution of 2-nitroaniline(15.0 g, 108 mmol) in dichloromethane (150 mL) was added dropwise to boron trifluoride diethyl etherate (23.1 g, 163 mmol) at -10°C to -15°C and stirred for 15 min, and a solution of isoamyl nitrite (15.26 g, 130 mmol) in dichloromethane (75 mL) was added dropwise at this temperature. At the end of addition, the reaction mixture was continuously stirred for 30 minutes, and then stirred for 30 minutes at -10°C to 0°C. The reaction system was added with cold petroleum ether (250 mL) dropwise and then filtered. The filter cake was washed with cold methyl tert-butyl ether (MTBE) (40 mL) to obtain 2-nitro-phenyl-diazonium tetrafluoroborate (1) (18.7 g). The yield was 73.1%.
  • Step B: a solution of potassium selenocyanate (8.0 g, 55.5 mmol) in water (80 mL) was added into a mixture of compound 1 (13.0 g, 54. 9 mmol) and water (300 mL) in an ice-water bath, and the reaction mixture was continuously stirred for 30 min at the end of addition. The reaction mixture was filtered, and the filter cake was washed with a small amount of water and then dried in vacuum at 60°C to obtain 1-nitro-2-phenyl selenocyanate (2) (11.2 g). The yield was 89.8%.
  • Step C: sodium (6.0 g, 261 mmol) was added to a mixture of compound 2 (10.5 g, 46.2 mol) and absolute ethyl alcohol (520 mL) at room temperature, and the mixture was stirred for 1 h in a water bath. The reaction mixture was cooled to 0°C to 5°C and filtered, the filter cake was washed with a small amount of cold ethanol, and the collected solid was suspended in methylbenzene (100 mL), heated to reflux to dissolve the product, and filtered immediately. The filtrate was cooled to 0°C to 5°C to separate out solid and then filtered, and the filter cake was collected to obtain 1,2-di(2-nitrophenyl)diselenide (3) (4.5 g). The yield was 48.4%.
  • Step D: zinc powder (13.5 g, 206 mmol) was added into a suspension solution of compound 3 (4.5 g, 11.2 mmol) in acetic acid (90 mL) at 40°C, and the reaction mixture was heated to 100°C and continuously stirred for 2 h. The reaction mixture was cooled below 50°C, 6M hydrochloric acid (40 mL) was slowly added dropwise and the mixture was filtered to remove insoluble substances. The filtrate was adjusted with 20% sodium acetate aqueous solution until the pH value was 2 to 3, and the solid was separated out. The reaction mixture was filtered, and the filter cake was dried to obtain di[(2-aminophenyl)seleno]zinc (4) (3.0 g). The yield was 77.8%.
  • Step E: a mixture of compound 4 (3.6 g, 17.4 mmol) and 4-nitrobenzoyl chloride (4.77 g, 25.7 mmol) was stirred for 2 h at 110°C. The mixture was cooled to room temperature, saturated sodium bicarbonate aqueous solution (60 mL) was added, the mixture was extracted with ethyl acetate (50 mL×3), and the combined organic phases were washed with saturated saline solution (20 mL) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:5 to 3:1) to obtain 2-(4-nitrophenyl)benzoselenazole (5) (500 mg). The yield was 6.42%. 1H NMR (DMSO-d 6, 400 MHz) δ 8.40 (dd, J = 2.0, 6.8 Hz, 2H), 8.21 (d, J = 8.8 Hz, 2H), 7.76-7.74 (m, 1H), 7.36-7.33 (m, 2H), 7.27-7.23 (m, 1H).
  • Step F: compound 5 (450 mg, 1.48 mmol) was dissolved into ethanol (10 mL), 2M hydrochloric acid (15 mL) and tin powder (2.25 g, 6.74 mmol) were then added, and the mixture was refluxed and stirred for 2 h. Most of the solvent was evaporated under reduced pressure, then water (15 mL) was added, and the mixture was adjusted with dilute sodium hydroxide solution until the pH value was 9 to 10, extracted with dichloromethane (20 mL×3), and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:15 to 1:4) to obtain 4-(benzoselenazole-2-yl)aniline (6) (300 mg). The yield was 74.2%. 1H NMR (DMSO-d 6, 400 MHz) δ 8.07 (dd, J = 0.8, 8.0 Hz, 1H), 7.89 (dd, J = 0.8, 8.0 Hz, 1H), 7.72-7.69 (m, 2H), 7.46-7.42 (m, 1H), 7.28-7.23 (m, 1H), 6.66-6.63 (m, 2H), 5.92 (s, 2H).
  • Step G: a solution of NBS (117 mg, 0.657 mmol) in dichloromethane (15 mL) was added dropwise into a solution of compound 6 (200 mg, 0.732 mmol) in dichloromethane (5 mL) at -10°C, and the mixture was continuously stirred for 0.5 h at this temperature at the end of addition. The reaction mixture was washed with water (10 mL) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:10) to obtain 4-(benzoselenazole-2-yl)-2-bromoaniline (7) (179 mg). The yield was 69.5%. 1H NMR (DMSO-d 6, 400 MHz) δ 8.10 (dd, J = 1.0, 8.0 Hz, 1H), 8.02 (d, J = 1.0 Hz, 1H), 7.94 (dd, J = 1.0, 8.0 Hz, 1H), 7.72 (dd, J = 1.0, 8.0 Hz, 1H), 7.49-7.45 (m, 1H), 7.31-7.27 (m, 1H), 6.88 (d, J = 8.4 Hz, 1H), 6.11 (s, 2H). MS (El, m/z):350.9 [M-H]-.
  • Embodiment 2: synthesis of 4-(5-fluorobenzoselenazole-2-yl)aniline (13) and 2-bromo-4-(5-fluorobenzoselenazole-2-yl)aniline (14)
  • Figure imgb0007
  • Step A: a solution of 2-nitro-4-fluoroaniline (5.0 g, 32.0 mmol) in dichloromethane (50 mL) was added dropwise to boron trifluoride diethyl etherate (6.8 g, 47.9 mmol) at -10°C to -15°C and stirred for 15 min, and a solution of isoamyl nitrite (4.5 g, 38.4 mmol) in dichloromethane (25 mL) was added dropwise at this temperature. At the end of addition, the mixture was continuously stirred for 30 minutes, and then stirred for 30 minutes at -10°C to 0°C. The reaction mixture was added with cooled petroleum ether (80 mL) dropwise and then filtered. The filter cake was washed with cold MTBE (10 mL) to obtain 2-nitro-4-fluorophenyl-diazonium tetrafluoroborate (8) (13.0 g). The compound was directly used in the next reaction without purification.
  • Step B: a solution of potassium selenocyanate (4.84 g, 33.6 mmol) in water (30 mL) was added to a mixture of crude compound 8 (13.0 g) and water (170 mL) in an ice-water bath, and the reaction mixture was continuously stirred for 20 min at the end of addition. The reaction mixture was filtered, and the filter cake was washed with a small amount of water and then dried in vacuum at 60°C to obtain 4-fluoro-2-nitro-1-phenyl selenocyanate (9) (9.1 g). The compound was directly used in the next reaction without purification.
  • Step C: sodium (4.1 g, 178 mmol) was added to a mixture of crude compound 9 (9.1 g) and absolute ethyl alcohol (300 mL) at room temperature, and the mixture was stirred for 1 h in a water bath. The mixture was cooled to 0°C to 5°C and filtered, and the filter cake was washed with a small amount of cold ethanol. The collected solid was suspended in methylbenzene (80 mL), heated to reflux to dissolve the product, and filtered immediately. The filtrate was cooled to 0°C to 5°C to separate out solid and then filtered, and the filter cake was collected to obtain 1,2-di(4-fluoro-2-nitrophenyl)diselenide (10) (2.0 g). The total yield of the reactions in the steps A, B and C was 21.9%.
  • Step D: compound 10 (1.49 g, 3.4 mmol) and Raney nickel (1.2 g) were suspended in isopropanol (30 mL), 85% hydrazine hydrate (1.8 mL) was added, and the mixture was heated to reflux and continuously stirred for 2.5 h. The mixture was immediately filtered through a celite pad, the filter cake was washed with a small amount of isopropanol, and the filtrate was collected. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:10 to 1:5) to obtain 6,6'-diselenodi(3-fluoroaniline) (11) (370 mg). The yield was 28.8%.
  • Step E: a mixture containing compound 11 (370 mg, 0.978 mmol), 4-nitrobenzaldehyde (296 mg, 1.958 mmol), sodium metabisulfite (372 mg, 1.956 mmol) and anhydrous DMSO (15 mL) was stirred for 72 h under nitrogen at 120°C. The mixture was cooled to room temperature, added with saturated ammonium chloride aqueous solution (60 mL), extracted with ethyl acetate (25 mL×3), and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified under reduced pressure by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:50) to obtain 5-fluoro-2-(4-nitrophenyl)benzoselenazole (12) (76 mg). The yield was 12.1%. 1H NMR (DMSO-d 6, 300 MHz) δ 8.39-8.28 (m, 5H), 8.01 (dd, J = 2.4, 9.9 Hz, 1H), 7.41-7.34 (m, 1H).
  • Step F: compound 12 (70 mg, 0.218 mmol) was dissolved into ethanol (5 mL) and then added with stannous chloride hydrate (246 mg, 1.09 mmol), and the mixture was refluxed and stirred for 5 h. The mixture was cooled to room temperature, saturated saline solution (20 mL) was added and the mixture was extracted with ethyl acetate (25 mL×3), and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:15 to 1:5) to obtain 4-(5-fluorobenzoselenazole-2-yl)aniline (13) (50 mg). The yield was 78.8%, MS (El, m/z):291.0 [M-H]-.
  • Step G: a solution of NBS (29.6 mg, 0.166 mmol) in dichloromethane (40 mL) was added dropwise to a solution of compound 13 (44 mg, 0.151 mmol) in dichloromethane (40 mL) in an ice-salt bath, and the mixture was continuously stirred for 10 min at this temperature at the end of addition. The reaction mixture was washed with water (20 mL×2) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:20) to obtain 2-bromo-4-(5-fluorobenzoselenazole-2-yl)aniline (14). 1H NMR (DMSO-d6, 300 MHz) δ 8.14-8.09 (m, 1H), 8.03 (d, J = 1.8 Hz, 1H), 7.78-7.70 (m, 2H), 7.23-7.16 (m, 1H), 6.87 (d, J = 8.4 Hz, 1H), 6.14 (s, 2H). MS (El, m/z):369.0 [M-H]-.
  • Embodiment 3: synthesis of 4-(5-fluorobenzoselenazole-2-yl)-2-methylaniline (17)
  • Figure imgb0008
  • Step A: zinc powder (5.5 g, 84.1 mmol) was added to a suspension of compound 10 (2.0 g, 4.57 mmol) in acetic acid (40 mL) at 40°C, then heated to 100°C and continuously stirred for 3 h. The reaction mixture was cooled below 50°C, 6M hydrochloric acid (40 mL) was slowly added and the mixture was filtered to remove insoluble substances. The filtrate was adjusted with 20% sodium acetate until the pH value was 2 to 3, and the solid was collected by filtration and dried to obtain di[(2-amino-4-fluorophenyl)seleno]zinc (15) (1.2 g). The yield was 61.9%.
  • Step B: a mixture of compound 15 (590 mg, 3.10 mmol) and 3-methyl-4-nitrobenzoyl chloride (663 mg, 3.32 mmol) was stirred for 4 h at 100°C. The mixture was cooled to room temperature, saturated sodium bicarbonate aqueous solution (15 mL) was added and the mixture was extracted with ethyl acetate (30 mL×2). The combined organic phases were washed with saturated saline solution (10 mL) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:15 to 1:1) to obtain 5-fluoro-2-(3-methyl-4-nitrophenyl)benzoselenazole (16) (750 mg). The yield was 72.2%. 1H NMR (DMSO-d 6, 300 MHz) δ 8.32-8.27 (m, 1H), 8.19-8.13 (m, 3H), 7.98 (dd, J = 2.4, 9.9 Hz, 1H), 7.40-7.33 (m, 1H), 2.64 (s, 3H).
  • Step C: compound 16 (200 mg, 0.597 mmol) was dissolved into ethanol (5 mL), 3M hydrochloric acid (4 mL) and tin powder (800 mg, 6.74 mmol) were then added and the mixture was refluxed and stirred for 1.5 h. Most of the solvent was evaporated under reduced pressure, water (15 mL) was then added and the mixture was adjusted with dilute sodium hydroxide solution until the pH value was 9 to 10, extracted with ethyl acetate (20 mL×2), and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:15 to 1:4) to obtain 4-(5-fluorobenzoselenazole-2-yl)-2-methylaniline (17) (103 mg). The yield was 56.4%. 1H NMR (DMSO-d 6, 400 MHz) δ 8.08-8.05 (m, 1H), 7.72-7.69 (m, 1H), 7.62-7.57 (m, 2H), 7.18-7.13 (m, 1H), 6.68 (d, J = 8.4 Hz, 1H), 5.75 (s, 2H), 2.14 (s, 3H). MS (El, m/z):305.0 [M-H]-.
  • Embodiment 4: synthesis of 2-bromo-4-(5-fluorobenzoselenazole-2-yl)-6-methylaniline (18)
  • Figure imgb0009
  • NBS (57 mg, 0.320 mmol) was added to a solution of compound 17 (88 mg, 0.229 mmol) in DMF (5 mL) at room temperature, and the mixture was continuously stirred for 20 min at this temperature at the end of addition. Water (25 mL) was added to the mixture which was then extracted with ethyl acetate (20 mL×2), and the combined organic phases were washed with saturated sodium bicarbonate aqueous solution (10 mL) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:5) to obtain 2-bromo-4-(5-fluorobenzoselenazole-2-yl)-6-methylaniline (18) (73 mg). The yield was 83.0%. 1H NMR (DMSO-d 6, 500 MHz) δ 8.13-8.10 (m, 1H), 7.92 (d, J = 2.0 Hz, 1H), 7.77-7.74 (m, 1H), 7.64 (s, 1H), 7.21-7.17 (m, 1H), 5.76 (s, 2H), 2.26 (s, 3H). MS (El, m/z):384.9 [M+H]+.
  • Embodiment 5: synthesis of 4-(5-bromobenzoselenazole-2-yl)-2-chloroaniline (25)
  • Figure imgb0010
  • Steps A, B and C referred to steps A, B and C in Embodiment 2.
  • Step D: compound 21 (1.9 g, 3.393 mmol) was dissolved into ethanol (40 mL) and stannous chloride hydrate (3.8 g, 16.84 mmol) was then added, and the mixture was refluxed and stirred for 3.5 h under nitrogen. The mixture was cooled to room temperature, saturated saline solution (20 mL) was added, the mixture was extracted with ethyl acetate (25 mL×3), and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:10) to obtain 6,6'-diselenodi(3-bromoaniline) (22) (450 mg). The yield was 26.5%.
  • Step E: referring to step E in Embodiment 2, to get 5-bromo-2-(4-nitrophenyl)benzoselenazole (23). 1H NMR (DMSO-d 6, 300 MHz) δ 8.39-8.30 (m, 5H), 8.23 (d, J = 9.0 Hz, 1H), 7.61 (d, J = 9.0 Hz, 1H).
  • Steps F and G: referring to step F in Embodiment 2 to get compound 24. The compound 23 was reduced to obtain a compound 24. Compound 24 (80 mg, 0.227 mmol) was dissolved into DMF (5 mL), NCS (34 mg, 0.255 mmol) was added and the mixture was stirred overnight at room temperature. Water (25 mL) was added to the mixture which was extracted with ethyl acetate (15 mL×3), and the combined organic phases were washed with saturated sodium bicarbonate aqueous solution (10 mL) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:15) to obtain 4-(5-bromobenzoselenazole-2-yl)-2-chloroaniline (25). 1H NMR (DMSO-d 6, 300 MHz) δ 8.10-8.04 (m, 2H), 7.87 (s, 1H), 7.68 (d, J = 9.0 Hz, 1H), 7.43 (d, J = 9.0 Hz, 1H), 6.88 (d, J = 9.0 Hz, 1H), 6.16 (s, 2H). MS (El, m/z):384.9 [M-H]-.
  • Embodiment 6: synthesis of 2-methyl-4-(5-methylbenzoselenazole-2-yl)aniline (26)
  • Figure imgb0011
  • The method for preparing compound 26 refers to steps A, B, C and D in Embodiment 2 and steps E and F in Embodiment 5. 2-Nitro-4-fluoroaniline in step A of Embodiment 2 was replaced with 2-nitro-4-methylaniline, and 4-nitrobenzaldehyde in step E of Embodiment 5 was replaced with 3-methyl-4-nitrobenzaldehyde. 1H NMR (DMSO-d 6, 500 MHz) δ 7.89 (d, J = 8.0 Hz, 1H), 7.71 (s, 1H), 7.59-7.54 (m, 2H), 7.08 (d, J = 8.0 Hz, 1H), 6.67 (d, J = 8.0 Hz, 1H), 5.62 (s, 2H), 2.41 (s, 3H), 2.14 (s, 3H). MS (El, m/z):301.0 [M-H]-.
  • Embodiment 7: synthesis of 2-methyl-4-[5-(trifluoromethyl)benzoselenazole-2-yl]aniline (27)
  • Figure imgb0012
  • The method for preparing compound 27 refers to steps A, B, C and D in Embodiment 2 and steps E and F in Embodiment 5. 2-nitro-4-fluoroaniline in step A of Embodiment 2 was replaced with 2-nitro-4-trifluoromethylaniline, and 4-nitrobenzaldehyde in step E of Embodiment 5 was replaced with 3-methyl-4-nitrobenzaldehyde. 1H NMR (DMSO-d 6, 500 MHz) δ 8.31 (d, J = 8.0 Hz, 1H), 8.16 (s, 1H), 7.65-7.62 (m, 2H), 7.55 (d, J = 8.5 Hz, 1H), 6.70 (d, J = 8.5 Hz, 1H), 5.77 (s, 2H), 2.15 (s, 3H). MS (El, m/z):355.0 [M-H]-.
  • Embodiment 8: synthesis of 2-(3,4-dimethoxy-phenyl)-5-fluoro-benzoselenazole (28)
  • Figure imgb0013
  • The method for preparing compound 28 refers to step B in Embodiment 3, wherein acyl chloride in the reaction equation was prepared by reacting a corresponding acid with thionyl chloride. MS (El, m/z):338.1 [M+H]+.
  • Embodiment 9: synthesis of 4-(6-ethoxybenzoselenazole-2-yl)-2-methylaniline (29)
  • Figure imgb0014
  • The method for preparing compound 29 refers to steps A, B, C and D in Embodiment 2 and steps E and F in Embodiment 5. 2-nitro-4-fluoroaniline in step A was replaced with 2-nitro-5-fluoroaniline, and 3-methyl-4-nitrobenzaldehyde in step E of Embodiment 5 was replaced with 3-methyl-4-nitrobenzaldehyde. In the reduction reactions in steps C and F, the solvent was ethanol so that F at the sixth site of benzaldehyde was substituted with ethoxy. The final resulting product was 4-(6-ethoxybenzoselenazole-2-yl)-2-methylaniline (29). 1H NMR (DMSO-d 6, 300 MHz) δ 7.76 (d, J = 8.7 Hz, 1H), 7.63 (s, 1H), 7.55-7.50 (m, 2H), 6.99 (d, J = 8.1 Hz, 1H), 6.66 (d, J = 8.1 Hz, 1H), 5.58 (s, 2H), 4.09 (q, J = 6.6 Hz, 2H), 2.13 (s, 3H), 1.35 (t, J = 6.6 Hz, 3H). MS (El, m/z):333.0 [M+H]+.
  • Embodiment 10: synthesis of 4-(6-ethoxy-5-fluorobenzoselenazole-2-yl)-2-methylaniline (30)
  • Figure imgb0015
  • The method for preparing compound 30 refers to steps A, B, C and D in Embodiment 2 and steps E and F in Embodiment 5. 2-nitro-4-fluoroaniline in step A of Embodiment 2 was replaced with 2-nitro-4,5-difluoroaniline, and 4-nitrobenzaldehyde in step E of Embodiment 5 was replaced with 3-methyl-4-nitrobenzaldehyde. In the reduction reactions in steps C and F, the solvent was ethanol so that F at the sixth site of benzaldehyde was substituted with ethoxy. The final resulting product was 4-(6-ethoxy-5-fluorobenzoselenazole-2-yl)-2-methylaniline (30). 1H NMR (DMSO-d 6, 300 MHz) δ 7.85 (d, J = 8.4 Hz, 1H), 7.72 (d, J = 12.0 Hz, 1H), 7.55-7.50 (m, 2H), 6.66 (d, J = 8.1 Hz, 1H), 5.61 (s, 2H), 4.16 (q, J = 6.9 Hz, 2H), 2.13 (s, 3H), 1.38 (t, J = 6.9 Hz, 3H). MS (El, m/z):351.0 [M+H]+.
  • Embodiment 11: synthesis of 5-(benzoselenazole-2-yl)-2-methoxyphenol (32)
  • Figure imgb0016
  • According to step D in Embodiment 5 and step E in Embodiment 2, compound 32 was synthesized as the aimed product while using compound 3 and 4-nitrobenzaldehyde as the starting materials.The compound 3 was used as raw material, and the method for preparing the compound 32 refers to step D in Embodiment 5 and step E in Embodiment 2. 4-nitrobenzaldehyde in step E of Embodiment 2 was replaced with 3-hydroxyl-4-methoxybenzaldehyde. 1H NMR (DMSO-d 6, 300 MHz) δ 9.55 (s, 1H), 8.13 (d, J = 7.5 Hz, 1H), 7.99 (d, J = 7.5 Hz, 1H), 7.51-7.44 (m, 3H), 7.36-7.30 (m, 1H), 7.06 (d, J = 8.4 Hz, 1H), 3.85 (s, 3H). MS (El, m/z):306.0 [M+H]+.
  • Embodiment 12: synthesis of 2-(3,4-dimethoxyphenyl)benzoselenazole (33)
  • Figure imgb0017
  • A mixture of compound 32 (90 mg, 0.296 mmol), potassium carbonate (61 mg, 0.441 mmol), iodomethane (126 mg, 0.888 mmol) and DMF (8 mL) was stirred overnight at 50°C. Water (40 mL) was added to the mixture which was extracted with ethyl acetate (20 mL×3), and the combined organic phases were washed with saturated saline solution (15 mL×2) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether=1:5) to obtain 2-(3,4-dimethoxy- phenyl)benzoselenazole (33). 1H NMR (DMSO-d 6, 300 MHz) δ 8.13 (d, J = 7.8 Hz, 1H), 8.02 (d, J = 7.8 Hz, 1H), 7.60-7.55 (m, 2H), 7.52-7.47 (m, 1H), 7.36-7.31 (m, 1H), 7.10 (d, J = 8.1 Hz, 1H), 3.89 (s, 3H), 3.85 (s, 3H). MS (EI, m/z):320.0 [M+H]+.
  • Embodiment 13: synthesis of 2-fluoro-4-(5-fluorobenzoselenazole-2-yl)aniline (34) and 2-bromo-6-fluoro-4-(5-fluorobenzoselenazole-2-yl)aniline (35)
  • Figure imgb0018
  • Step A: tributylphosphine (1.28 g, 6.33 mmol) was added to a solution of compound 11 (800 mg, 2.12 mmol) in methylbenzene (15 mL), then stirred for 5 min under nitrogen. To the mixture was added 3-fluoro-4-nitrobenzoic acid (392 mg, 2.12 mmol), and the resulting mixture was refluxed for 48 h under nitrogen. The mixture was cooled to room temperature, water (25 mL) was added and the mixture was adjusted with saturated sodium carbonate solution until the pH value was 9 to 10. The mixture was extracted with ethyl acetate (20 mL×3), and the combined organic phases were washed with saturated saline solution (15 mL) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:15) to obtain 2-fluoro-4-(5-fluoro-benzoselenazole-2-yl)aniline (34). 1H NMR (DMSO-d 6, 300 MHz) δ 8.13-8.09 (m, 1H), 7.77-7.73 (m, 1H), 7.69-7.64 (m, 1H), 7.59-7.56 (m, 1H), 7.22-7.16 (m, 1H), 6.88-6.83 (m, 1H), 6.00 (s, 2H). MS (El, m/z):309.0 [M-H]-.
  • Step B: 2-bromo-6-fluoro-(5-fluorobenzoselenazole-2-yl)aniline (35) was prepared by using compound 34 as raw material, and the method refers to step G in Embodiment 2. 1H NMR (DMSO-d 6, 500 MHz) δ 8.16-8.13 (m, 1H), 7.90 (s, 1H), 7.79 (dd, J = 2.0, 10.0 Hz, 1H), 7.73 (dd, J = 2.0, 10.0 Hz, 1H), 7.24-7.20 (m, 1H), 6.12 (s, 2H). MS (El, m/z):388.9 [M+H]+.
  • Embodiment 14: synthesis of 5-(5-fluorobenzoselenazole-2-yl)-2-methylaniline (37)
  • Figure imgb0019
  • Step A: a solution of compound 11 (800 mg, 2.12 mmol) and tributylphosphine (1.28 g, 6.32 mmol) in methylbenzene (15 mL) was stirred for 5 min under nitrogen, 3-nitro-4-methylbenzoic acid (380 mg, 2.09 mmol) was added, and the mixture was refluxed and stirred for 48 h under nitrogen. The mixture was cooled to room temperature, water (30 mL) was added and the mixture was adjusted with 2M sodium hydroxide solution until the pH value was 9 to 10. The mixture was extracted with ethyl acetate (20 mL×3) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:20 to 1:2) to obtain 5-fluoro-2-(4-methyl-3-nitrophenyl)benzoselenazole (36) (107 mg). The yield was 15.3%.
  • Step B: 5-(5-fluorobenzoselenazole-2-yl)-2-methylaniline (37) was prepared by using compound 36 as raw material, and the experimental operation refers to step F in Embodiment 2. 1H NMR (DMSO-d 6, 500 MHz) δ 8.17-8.14 (m, 1H), 7.82 (dd, J = 2.5, 10.0 Hz, 1H), 7.38 (s, 1H), 7.27-7.23 (m, 1H), 7.18 (d, J = 8.0 Hz, 1H), 7.10 (d, J = 8.0 Hz, 1H), 2.13 (s, 3H). MS (El, m/z):305.1 [M-H]-.
  • Embodiment 15: synthesis of 2-[3-chloro-4-(trifluoromethoxy)phenyl)-5-fluorobenzoselenazole (38)
  • Figure imgb0020
  • The method for preparing compound 38 refers to step E in Embodiment 2, wherein 4-nitrobenzaldehyde in step E of Embodiment 2 was replaced with 3-chloro-4(trifluoromethoxy)benzaldehyde. 1H NMR (DMSO-d 6, 300 MHz) δ 8.35 (d, J = 1.2 Hz, 1H), 8.30-8.27 (m, 1H), 8.17-8.15 (m, 1H), 7.98-7.96 (m, 1H), 7.77-7.75 (m, 1H), 7.38-7.34 (m, 1H). MS (El, m/z):394.0 [M-H]-.
  • Embodiment 16: synthesis of 4-(5-deuterobenzoselenazole-2-yl)-2-methylaniline (40)
  • Figure imgb0021
  • Step A: compound 22 and 3-methyl-4-nitrobenzaldehyde were used as raw materials, and the experimental operation of preparing compound 39 refers to step E in Embodiment 2. 1H NMR (DMSO-d 6, 400 MHz) δ 8.35 (s, 1H), 8.24 (d, J = 8.4 Hz, 1H), 8.19 (s, 1H), 8.15-8.14 (m, 2H), 7.62 (dd, J = 2.0, 8.4 Hz, 1H), 2.65 (s, 3H).
  • Step B: compound 39 (42 mg, 0.106 mmol) was suspended in DMF (5 mL) and deuteroxide (0.5 mL) and 5% palladium carbon were then added, and the mixture was stirred overnight under deuterium gas under normal pressure. The mixture was filtered through a celite pad, water (20 mL) was added and the mixture was extracted with ethyl acetate (20 mL×2), and the combined organic phases were washed with saturated saline solution (10 mL×2) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:10) to obtain 4-(5-deuterobenzoselenazole-2-yl)-2-methylaniline (40). 1H NMR (DMSO-d 6, 400 MHz) δ 8.06-8.02 (m, 2H), 7.62-7.59 (m, 2H), 8.16 (dd, J = 2.0, 8.8 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 5.78 (s, 2H), 2.14 (s, 3H). MS (El, m/z):288.0 [M-H]-.
  • Embodiment17: synthesis of 2,6-difluoro-4-(5-fluorobenzoselenazole-2-yl)aniline (41)
  • Figure imgb0022
  • The method for preparing compound 41 refers to step A in Embodiment 13, wherein 3-fluoro-4-nitrobenzaldehyde in step A of Embodiment 13 was replaced with 3,5-difluoro-4-nitrobenzaldehyde. 1H NMR (DMSO-d 6, 400 MHz) δ 8.18-8.14 (m, 1H), 7.79 (dd, J = 2.4, 10.0 Hz, 1H), 7.62 (dd, J = 2.4, 7.2 Hz, 2H), 7.26-7.21 (m, 1H), 6.11 (s, 2H). MS (El, m/z):327.0 [M-H]-.
  • Embodiment 18: synthesis of 2-fluoro-4-(5-fluorobenzoselenazole-2-yl)-6-methylaniline (45)
  • Figure imgb0023
  • Step A: a solution of NBS (5.0 g, 28.1 mmol) in DMF (20 mL) was added dropwise to a solution of 2-fluoro-6-methylaniline (3.5 g, 28.0 mmol) in DMF (10 mL) in an ice-water bath, and the mixture was continuously stirred for 5 min at the end of addition. The ice-water bath was removed, and the reaction mixture was stirred 0.5 h at room temperature. Water (150 mL) was added to the mixture which was extracted with ethyl acetate (80 mL×3), and the combined organic phases were successively washed with saturated sodium bicarbonate solution (40 mL×2) and saturated saline solution (40 mL×2), and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure to obtain 4-bromo-2-fluoro-methylaniline (42) (5.3 g). The yield was 92.8%.
  • Step B: a mixture of compound 42 (5.3 g, 26.0 mmol), cuprous cyanide (3.0 g, 33.5 mmol) and N-methylpyrrolidone (15 mL) was stirred overnight under nitrogen at 180°C. Water (75 mL) was added to the mixture which was extracted with ethyl acetate (50 mL×3), and the combined organic phases were successively washed with water (30 mL×2) and saturated saline solution (30 mL) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:30 to 1:5) to obtain 4-amino-3-fluoro-5-methylbenzonitrile (43) (2.96 g). The yield was 75.8%.
  • Step C: a mixture of compound 43 (2.95 g, 19.6 mmol), 1M sodium hydroxide solution (50 mL) and ethanol (5 mL) was refluxed and stirred overnight. The mixture was cooled to room temperature, water (50 mL) was added and the mixture was washed with MTBE (20 mL×2), and the water phase was used as the product. The water phase was adjusted with 2M hydrochloric acid until the pH value was 3 to 4, and the solid was separated out. The mixture was filtered, and the filter cake was dried to obtain 4-amino-3-fluoro-5-methylbenzoic acid (44) (2.90 g). The yield was 87.5%.
  • Step D: tributylphosphine (2.15 g, 10.6 mmol) was added to a mixture of compound 11 (1.74 g, 4.60 mmol), compound 44 (600 mg, 3.55 mmol) and methylbenzene (25 mL), and the mixture was refluxed and stirred for 48 h under nitrogen. The mixture was cooled to room temperature, water (40 mL) was added and the mixture was adjusted with 2M sodium hydroxide solution until the pH value was 9 to 10. The mixture was extracted with ethyl acetate (40 mL×3), and the combined organic phases were washed with saturated saline solution (25 mL) and dried over anhydrous sodium sulfate. The solvent was evaporated under reduced pressure, and the product was purified by chromatography (200 to 300 meshes of silica gel, elution with ethyl acetate: petroleum ether = 1:100 to 1:30) to obtain 2-fluoro-4-(5-fluorobenzoselenazole-2-yl)-6-methylaniline (45). 1H NMR (DMSO-d6, 400 MHz) δ 8.14-8.10 (m, 1H), 7.77-7.74 (m, 1H), 7.58-7.55 (m, 1H), 7.51 (s, 1H), 7.22-7.17 (m, 1H), 5.76 (s, 2H), 2.22 (s, 3H). MS (El, m/z):323.0 [M-H]-.
  • Bioactivity Embodiments Embodiment 19: Inhibition effects of the compounds on the growth of breast cancer cell lines of MCF-7 and MDA-MB-468 Experimental methods and results
    1. 1. Breast cancer cells MCF-7 (Luminal type cells) and MDA-MB-468 (basal-like cells, which are of the triple-negative type without epithelial-mesenchymal transition) were purchased from Cell Resource Center of Shanghai Institutes for Biological Sciences of the Chinese Academy of Sciences, and cultured with DMEM culture medium (containing 10% of fetal bovine serum, 10 U/mL of penicillin and 0.1 mg/mL of streptomycin) in a 5% CO2 incubator at 37°C until the cell density was about 90%.
    2. 2. The cells were inoculated to a 96-well plate at a cell population of 3×103/well and then cultured in the 5% CO2 incubator for 24 h at 37°C.
    3. 3. Tested compounds of different concentration gradients were prepared by using the DMEM culture medium, and added into wells at 100 µL/well as tested compounds wells; and, the DMEM culture liquid was added into wells at 100 µL/well as negative control wells. At 37°C and in the 5% CO2 incubator, the MCF-7 cells were cultured for 120 h, and MDA-MB-468 cells were cultured for 72 h.
    4. 4. Resazurin (15 mg/50 mL), Methylene Blue (25 mg/10 mL), Potassium ferricyanide (0.329 g/100 mL) and Potassium ferrocyanide (0.422 g/100 mL) were dissolved into PBS (0.1 M, pH=7.4) to obtain Alamar Blue solution for standby.
    5. 5. The cells were washed twice with PBS (0.1 M, pH=7.4), and the Alamar Blue solution was added into wells at 100 µL/well; 100 µL of Alamar Blue solution was added into wells without cells to serve as blank control cells. The 96-well plate was placed into the 5% CO2 incubator at 37°C and cultured for 3 h.
    6. 6. The fluorescence value of the cells was detected at 530/590 nm by ELIASA Victor X4 (Perkin Elmer). The fluorescence value at each concentration was repetitively measured 4 times to obtain an average value and a standard deviation. The cell viability was calculated by the following formula: Cell viability % = tested compound wells blank control wells negative control wells blank control wells × 100 %
      Figure imgb0024
    7. 7. The half inhibitory concentration (IC50) of the tested compounds against the cell lines was obtained according to the cell viability by Prism Graph software. The experimental results are shown in Table 1.
    Table 1: Half inhibitory concentration (IC50, nM) of the compounds against cell lines of MCF-7 and MDA-MB-468
    Compound ID MCF-7 MDA-MB-468
    7 56.69 74.07
    14 187.71 137.94
    17 72.81 58.45
    18 91.86 70.93
    26 133.8
    34 52.38 29.06
    35 41.59 39.67
    37 449.56 113.63
    41 56.77 20.1
    Paclitaxel 4.6* 4.43
    *Note: In Table 1, the positive control drug paclitaxel has only 70% inhibition rate to MCF-7 cell line at its highest concentration at 500 nM. Therefore, the IC50 of the paclitaxel for the MCF-7 cell strains calculated by the software is lower.
    Embodiment 20: Inhibition effects of the compounds on the growth of human lung cancer cells H1299, human colon cancer cells HT29, human liver cancer cells SK-HEP-1, human colon cancer cells HCT116, human normal liver cells L-02 and WRL-68
  • Growth inhibition tests of the tested compounds 7, 17, 18, 34, 35 and 41 on the human lung cancer cells H1299, human colon cancer cells HT29, human liver cancer cells SK-HEP-1, human colon cancer cells HCT116, human normal liver cells L-02 and WRL-68 were conducted, and the method refers to Embodiment 19 "Inhibition effects of the compounds on the growth of breast cancer cells MCF-7 and MDA-MB-468". Compounds 7, 17, 18, 34, 35 and 41 had a half inhibitory concentration (IC50) greater than 10 µM for the cells, and had no obvious inhibition effects. The half inhibitory concentration (IC50) of the positive control drug paclitaxel for the six cell lines was within a range of 1.59 nM to 15.31 nM.
  • The experimental results indicated that the compounds 7, 17, 18, 34, 35 and 41 had very good growth inhibition effects on the breast cancer cells MCF-7 and MDA-MB-468; and the half inhibitory concentration (IC50) for other tested cell strains such as H1299, HT29, SK-HEP-1, HCT116, L-02 and WRL-68 was greater than 10 µM; and, the positive medicine paclitaxel still had strong inhibition toxicity on cancer cells in addition to the breast cancer cells and normal cells. Therefore, it was indicated that the compounds of the present application had remarkable selectivity for the inhibition of breast cancer cell lines.

Claims (10)

  1. A compound of formula (I), or a pharmaceutically acceptable salt thereof or a hydrolysable prodrug thereof:
    Figure imgb0025
    wherein:
    R1 and R2 are independently selected from H, D, halogen, -CN, C1-3 alkyl, substituted C1-3 alkyl, C1-3 alkoxy or substituted C1-3 alkoxy, and the substituent group is selected from D, halogen or C1-3 alkoxy;
    R3 and R4 are independently selected from H, D, halogen, -OH, -CN, -NH2, substituted -NH2, C1-3 alkyl, substituted C1-3 alkyl, C1-3 alkoxy or substituted C1-3 alkoxy, and the substituent group is selected from D, halogen, C1-3 alkyl or C1-3 alkoxy; and
    R5 is selected from H, -OH, -NH2, C1-3 alkyl, substituted C1-3 alkyl, C1-3 alkoxy or substituted C1-3 alkoxy, and the substituent group is selected from D, halogen,-OH, -NH2 or C1-3 alkoxy.
  2. The compound, pharmaceutically acceptable salt thereof or hydrolysable prodrug thereof according to claim 1, wherein:
    R1 and R2 are independently selected from H, D, halogen, -CN, C1-3 alkyl, substituted C1-3 alkyl, C1-3 alkoxy or substituted C1-3 alkoxy, and the substituent group is selected from D, F or C1-3 alkoxy;
    R3 and R4 are independently selected from H, D, halogen, -OH, -CN, -NH2, substituted -NH2, C1-3 alkyl, substituted C1-3 alkyl, C1-3 alkoxy or substituted C1-3 alkoxy, and the substituent group is selected from D, F, C1-3 alkyl or C1-3 alkoxy; and
    R5 is selected from H, -OH, -NH2, C1-3 alkoxy or substituted C1-3 alkoxy, and the substituent group is selected from D, F, -OH, -NH2 or C1-3 alkoxy.
  3. The compound, pharmaceutically acceptable salt thereof or hydrolysable prodrug thereof according to claim 1, wherein R1 and R2 are independently selected from H, D, F, Cl, -CN, -CH3, -CF3, -OCH3, -OCH2CH3, -OCF3 or -OCHF2.
  4. The compound, pharmaceutically acceptable salt thereof or hydrolysable prodrug thereof according to claim 1, wherein R3 and R4 are independently selected from H, D, halogen, -OH, -CN, -NH2, -CH3, -CH2CH3, -CF3, -OCH3,-OCH2CH3, -OCHF2 or -OCF3.
  5. The compound, pharmaceutically acceptable salts thereof or hydrolysable prodrugs thereof according to claim 1, wherein R5 is selected from H, -NH2, -CH3,-CF3, -OCH3, -OCHF2, -OCF3, -OCH2CH3 or -OCH2CF3.
  6. The compound, pharmaceutically acceptable salt thereof or hydrolysable prodrug thereof according to claim 1, wherein the compound is a compound of formula (II):
    Figure imgb0026
  7. The compound, pharmaceutically acceptable salt thereof or hydrolysable prodrug thereof according to claim 6, wherein R1 and R2 are independently selected from H, D, F, Cl, -CN, -CH3, -CF3 or -CHF2; and, R3 and R4 are independently selected from H, D, F, Cl, Br, I, -CN, -CH3, -CF3, -OCH3, -OCH2CH3, -OCHF2 or -OCF3.
  8. The compound, pharmaceutically acceptable salt thereof or hydrolysable prodrug thereof according to one of claims 1 to 7, wherein the compound is selected from:
    4-(benzoselenazole-2-yl)-2-bromoaniline,
    2-bromo-4-(5-fluorobenzoselenazole-2-yl)aniline,
    4-(5-fluorobenzoselenazole-2-yl)-2-methylaniline,
    4-(5-fluorobenzoselenazole-2-yl)aniline,
    2-bromo-4-(5-fluorobenzoselenazole-2-yl)-6-methylaniline,
    4-(5-bromobenzoselenazole-2-yl)-2-chloroaniline,
    2-methyl-4-(5-methylbenzoselenazole-2-yl)aniline,
    2-methyl-4-[5-(trifluoromethyl)benzoselenazole-2-yl]aniline,
    2-(3,4-dimethoxy-phenyl)-5-fluoro-benzoselenazole,
    4-(6-ethoxybenzoselenazole-2-yl)-2-methylaniline,
    4-(6-ethoxy-5-fluorobenzoselenazole-2-yl)-2-methylaniline,
    5-(benzoselenazole-2-yl)-2-methoxyphenol,
    2-(3,4-dimethoxyphenyl)benzoselenazole,
    2-fluoro-4-(5-fluorobenzoselenazole-2-yl)aniline,
    2-bromo-6-fluoro-4-(5-fluorobenzoselenazole-2-yl)aniline,
    5-(5-fluorobenzoselenazole-2-yl)-2-methylaniline,
    2-[3-chloro-4-(trifluoromethoxy)phenyl]-5-fluorobenzoselenazole,
    4-(5-deuterobenzoselenazole-2-yl)-2-methylaniline,
    2,6-difluoro-4-(5-fluorobenzoselenazole-2-yl)aniline, and
    2-fluoro-4-(5-fluorobenzoselenazole-2-yl)-6-methylaniline.
  9. A pharmaceutical composition containing a compound, pharmaceutically acceptable salt thereof or hydrolysable prodrug thereof according to one of claims 1 to 7 as active component or main active component, supplemented by pharmaceutically acceptable adjuvants.
  10. An application of the compound, pharmaceutically acceptable salt thereof or hydrolysable prodrug thereof according to one of claims 1 to 7 in preparing medicines for treating or preventing breast cancer.
EP16792124.6A 2015-05-09 2016-05-06 Compound for treating or preventing breast cancer Active EP3296294B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PL16792124T PL3296294T3 (en) 2015-05-09 2016-05-06 Compound for treating or preventing breast cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201510239715 2015-05-09
PCT/CN2016/081241 WO2016180274A1 (en) 2015-05-09 2016-05-06 Compound for treating or preventing breast cancer

Publications (3)

Publication Number Publication Date
EP3296294A1 true EP3296294A1 (en) 2018-03-21
EP3296294A4 EP3296294A4 (en) 2018-11-07
EP3296294B1 EP3296294B1 (en) 2020-02-12

Family

ID=56913857

Family Applications (1)

Application Number Title Priority Date Filing Date
EP16792124.6A Active EP3296294B1 (en) 2015-05-09 2016-05-06 Compound for treating or preventing breast cancer

Country Status (8)

Country Link
US (1) US10005744B2 (en)
EP (1) EP3296294B1 (en)
JP (2) JP6618550B2 (en)
CN (1) CN105949149B (en)
ES (1) ES2779465T3 (en)
PL (1) PL3296294T3 (en)
PT (1) PT3296294T (en)
WO (1) WO2016180274A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6618550B2 (en) * 2015-05-09 2019-12-11 江▲蘇▼新元素医▲薬▼科技有限公司 Compounds for the prevention or treatment of breast cancer
CN111170962B (en) * 2018-11-12 2023-05-23 江苏新元素医药科技有限公司 Use of 4- (benzoselenazol-2-yl) arylamine compounds for treating intestinal cancer
CN111170963B (en) * 2018-11-12 2023-05-23 江苏新元素医药科技有限公司 Use of 4- (benzoselenazol-2-yl) arylamine compound for treating gastric cancer
WO2020098517A1 (en) * 2018-11-12 2020-05-22 江苏新元素医药科技有限公司 Use of 4-(benzselenazol-2-yl)arylamine compound in treating stomach cancer or intestinal cancer

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5861424A (en) * 1991-04-26 1999-01-19 Dana Farber Cancer Institute Composition and method for treating cancer
GB9317949D0 (en) * 1993-08-28 1993-10-13 Stevens Malcolm F G Benzothiazole compounds
US6110929A (en) * 1998-07-28 2000-08-29 3M Innovative Properties Company Oxazolo, thiazolo and selenazolo [4,5-c]-quinolin-4-amines and analogs thereof
GB9919673D0 (en) * 1999-08-20 1999-10-20 Cancer Res Campaign Tech 2-Arlybenzazole compounds
EP2193127A4 (en) * 2007-09-27 2011-09-14 Inst Medical W & E Hall Benzothiazole compounds
CN102617478B (en) * 2011-01-31 2015-07-15 华东理工大学 Synthesis of benzimidazole, oxazole and thiazole derivatives and application thereof
WO2015051188A1 (en) * 2013-10-02 2015-04-09 Washington University Heterocyclic molecules for biomedical imaging and therapeutic applications
JP6618550B2 (en) * 2015-05-09 2019-12-11 江▲蘇▼新元素医▲薬▼科技有限公司 Compounds for the prevention or treatment of breast cancer

Also Published As

Publication number Publication date
EP3296294A4 (en) 2018-11-07
PL3296294T3 (en) 2020-08-24
EP3296294B1 (en) 2020-02-12
CN105949149B (en) 2018-08-24
JP2019196362A (en) 2019-11-14
WO2016180274A1 (en) 2016-11-17
JP2018515540A (en) 2018-06-14
US20180134673A1 (en) 2018-05-17
PT3296294T (en) 2020-05-13
JP6974869B2 (en) 2021-12-01
ES2779465T3 (en) 2020-08-17
US10005744B2 (en) 2018-06-26
CN105949149A (en) 2016-09-21
JP6618550B2 (en) 2019-12-11

Similar Documents

Publication Publication Date Title
JP6974869B2 (en) Compounds for the prevention or treatment of breast cancer
JP5662564B2 (en) Arylaminopurine derivatives, methods for their preparation and use as pharmaceuticals
EP3196197A1 (en) Indoleamine-2,3-dioxygenase inhibitor and preparation method therefor
JP2011527291A (en) Icotinib hydrochloride, compound, crystallographic form, concomitant drug and its use
EP2906564B1 (en) Treating brain cancer using agelastatin a (aa) and analogues thereof
WO2018149231A1 (en) 7h-pyrrolo[2,3-d]pyrimidine derivative and anti-tumour use thereof
JP6606806B2 (en) Deuterated quinazolinone compound and drug composition containing the compound
EA032032B1 (en) Derivatives of 2-aminopyridine as adenosine areceptor antagonists and ligands of melatonin mtreceptors
US9399644B2 (en) [1,3] dioxolo [4,5-G] quinoline-6(5H)thione derivatives as inhibitors of the late SV40 factor (LSF) for use in treating cancer
CN114507190B (en) Deuterated 1, 4-benzodiazepine-2, 5-diketone compound and application thereof
EP4098647A1 (en) Disubstituted adamantyl derivative or pharmaceutically acceptable salt thereof, and pharmaceutical composition for suppressing cancer growth comprising same as active ingredient
JP7184302B2 (en) Compounds Useful for Inhibiting Human Trefoil Factor 3
CN111170962B (en) Use of 4- (benzoselenazol-2-yl) arylamine compounds for treating intestinal cancer
RU2815432C2 (en) Composition used to inhibit human trefoil factor 3
CN111617085B (en) Targeted HDAC inhibitor and application thereof in antitumor therapeutic drugs
WO2020098516A1 (en) Use of 4-(benzothiazole-2-yl)arylamine compound in treating stomach cancer
EP3596054B1 (en) 5-carboxamide-2-thiobarbituric acids and use thereof as medicaments
KR20220104734A (en) Oleanane cinnamamide derivatives, preparation method and use thereof
CN116251107A (en) Application of Z-2-366 in preparation of medicines for inhibiting eEF2K expression
JP6013451B2 (en) Asymmetrically substituted anthrapyridazone derivatives as cytostatics
JP2019089720A (en) Compound or pharmaceutically acceptable salt thereof, myogenesis promoter, muscular atrophy inhibitor, pharmaceutical composition, and taz activator

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20171207

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20181008

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/41 20060101ALI20181001BHEP

Ipc: C07D 293/12 20060101AFI20181001BHEP

Ipc: A61P 35/00 20060101ALI20181001BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20190925

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1231942

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200215

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602016029670

Country of ref document: DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: BOVARD AG PATENT- UND MARKENANWAELTE, CH

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 3296294

Country of ref document: PT

Date of ref document: 20200513

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20200505

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200512

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2779465

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20200817

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200612

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200513

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200512

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602016029670

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1231942

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200212

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20201113

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200212

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602016029670

Country of ref document: DE

Representative=s name: CBDL PATENTANWAELTE GBR, DE

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20230505

Year of fee payment: 8

Ref country code: NL

Payment date: 20230525

Year of fee payment: 8

Ref country code: IT

Payment date: 20230510

Year of fee payment: 8

Ref country code: FR

Payment date: 20230428

Year of fee payment: 8

Ref country code: ES

Payment date: 20230609

Year of fee payment: 8

Ref country code: DE

Payment date: 20230613

Year of fee payment: 8

Ref country code: CH

Payment date: 20230701

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230504

Year of fee payment: 8

Ref country code: SE

Payment date: 20230509

Year of fee payment: 8

Ref country code: PL

Payment date: 20230428

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20230526

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230620

Year of fee payment: 8